US11278569B2 - Natural killer cell containing exogenous mitochondrium and pharmaceutical composition comprising same - Google Patents

Natural killer cell containing exogenous mitochondrium and pharmaceutical composition comprising same Download PDF

Info

Publication number
US11278569B2
US11278569B2 US16/349,310 US201716349310A US11278569B2 US 11278569 B2 US11278569 B2 US 11278569B2 US 201716349310 A US201716349310 A US 201716349310A US 11278569 B2 US11278569 B2 US 11278569B2
Authority
US
United States
Prior art keywords
cells
cancer
natural killer
mitochondria
killer cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US16/349,310
Other languages
English (en)
Other versions
US20190269731A1 (en
Inventor
Kyuboem Han
Youngjun Lee
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Paean Biotechnology Inc
Original Assignee
Paean Biotechnology Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Paean Biotechnology Inc filed Critical Paean Biotechnology Inc
Assigned to PAEAN BIOTECHNOLOGY INC. reassignment PAEAN BIOTECHNOLOGY INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HAN, KYUBOEM, LEE, YOUNGJUN
Publication of US20190269731A1 publication Critical patent/US20190269731A1/en
Application granted granted Critical
Publication of US11278569B2 publication Critical patent/US11278569B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions

Definitions

  • the present invention relates to a cell therapy products, and more particularly, to a natural killer (hereinafter referred to as NK) cell or a peripheral blood mononuclear cells (hereinafter referred to as PBMCs) which comprise exogenous mitochondria, and pharmaceutical compositions comprising the same as an active ingredient.
  • NK natural killer
  • PBMCs peripheral blood mononuclear cells
  • the biopharmaceuticals include protein drugs, antibody drugs and cell therapy products.
  • the cell therapy products refer to the pharmaceuticals used for the purpose of treatment, diagnosis, or prevention of diseases and the cell therapy products may be obtained through a series of actions such as by performing ex vivo isolation and proliferation or by altering biological characteristics of cells by other methods.
  • the cell therapy products may come from autologous, allogeneic, or xenogeneic cells. Depending on the cell type, the cell therapy products can be categorized into somatic cell therapy products and stem cell therapy products.
  • a PBMC in human blood is a blood cell with a round nucleus such as a lymphocyte or a monocyte, and includes B cells, T cells, macrophages, dendritic cells (DCs), and NK cells.
  • B cells B cells
  • T cells T cells
  • macrophages macrophages
  • DCs dendritic cells
  • NK and cytotoxic T lymphocytes (CTLs) cells directly eliminate cancer cells.
  • Antigen-presenting cells that present antigens to these effector cells include dendritic cells or B cells.
  • helper T cells and regulatory T cells which secrete various cytokines, and the like are involved in immune responses.
  • NK cells are emerging and promising immune cells in immune cell therapies.
  • NK cells have been shown to have an ability to kill cancer cells in a non-specific manner
  • many studies have been conducted on NK cells.
  • NK cell therapies in cancer have been emerging tools for treatment of cancer cells.
  • NK cells play an important role in innate and acquired immune responses achieved through cytokine secretion, against pathogens that have infected a host or cancer.
  • the present inventors have made efforts to find a new method for improving the power of cytotoxicity of NK cells and PBMCs. As a result, the present inventors have found a method of increasing the cytotoxicity of NK and PBMC, and thus provide a method of treating cancer using such cytotoxicity, and thus have completed the present invention.
  • An object of the present invention is to provide an NK cell with increased cytotoxicity and a pharmaceutical composition comprising the same.
  • Another object of the present invention is to provide a PBMC with increased cytotoxicity and a pharmaceutical composition comprising the same.
  • the present invention provides NK cells comprising exogenous mitochondria, and a pharmaceutical composition for preventing or treating cancer or an infectious disease, comprising the same.
  • the present invention provides PBMCs comprising exogenous mitochondria, and a pharmaceutical composition for preventing or treating cancer or an infectious disease, comprising the same.
  • NK cell and a PBMC into which exogenous mitochondria have been introduced, not only have an increased cytotoxicity which results in increased cancer-specific killing effects, but also exhibit no side effects as immune cells existing in vivo.
  • the NK cell and the PBMC are improved in terms of an ability of the cell itself, and thus can be widely applied to various diseases in which the NK cell and the PBMC are involved.
  • pharmaceutical compositions comprising the NK cell and the PBMC are highly commercially applicable.
  • FIG. 1 illustrates a result obtained by a PCR analysis method in order to investigate whether human normal hepatocyte (WRL68)-derived mitochondria have been delivered into NK cells.
  • FIG. 2 illustrates a result obtained by FACS analysis in order to investigate whether human normal hepatocyte-derived mitochondria have been delivered into NK cells.
  • FIG. 3 illustrates results obtained by fluorescence microscopy, in order to locate the human normal hepatocyte-derived mitochondria delivered into NK cells.
  • FIGS. 4A and 4B illustrate results showing anti-cancer activity of NK cells into which exogenous mitochondria have been delivered by a CD107a degranulation assay.
  • FIG. 5 illustrates a result showing the cytotoxic effects of NK cells into which exogenous mitochondria have been delivered by K562 cytotoxicity assay.
  • FIG. 6 illustrates a result obtained by FACS analysis in order to investigate whether mitochondria derived from umbilical cord mesenchymal stem cells (UC-MSC) have been delivered into NK cells.
  • UC-MSC umbilical cord mesenchymal stem cells
  • FIG. 7 illustrates a result showing the cytotoxic effects of NK cells into which UC-MSC-derived mitochondria have been delivered using K562 cytotoxicity assay.
  • FIGS. 8A to 8C illustrate results showing the therapeutic effects by NK cells, into which UC-MSC-derived mitochondria have been delivered in an animal model of acute myelogenous leukemia, in terms of body weight and survival rate of mice.
  • FIG. 9 illustrates results of expression distribution of blood tumor markers in an animal model of acute myelogenous leukemia which has been administered with NK cells into which UC-MSC-derived mitochondria have been delivered.
  • FIG. 10 illustrates a result showing the cytotoxic effects of PBMCs into which exogenous mitochondria have been delivered by K562 cytotoxicity assay
  • an NK cell enriched by exogenous mitochondria.
  • the term “exogenous mitochondria” refers to mitochondria introduced exogenously rather than mitochondria present in an NK cell.
  • the exogenous mitochondria may be obtained from the same subject as that from which the NK cell is obtained, but may be obtained from another subject.
  • the exogenous mitochondria may be obtained from a mammal, and preferably may be obtained from a human.
  • the exogenous mitochondria may be obtained from muscle cells, hepatocytes, fibroblasts, epithelial cells, neurons, adipocytes, osteocytes, leukocytes, lymphocytes, or mucosal cells, and preferably may be obtained from muscle cells with excellent mitochondrial activity.
  • the mitochondria may be obtained from cells cultured ex vivo.
  • the exogenous mitochondria can be obtained by disrupting the cells and performing centrifugation, or by culturing the cells, disrupting the cells, and performing the centrifugation.
  • a conventional method used for collecting an organelle can be used.
  • an NK cell comprising exogenous mitochondria may be obtained by introducing mitochondria in an amount of 0.01 to 500 ⁇ g, 0.1 to 450 ⁇ g, 0.5 to 300 ⁇ g, 1 to 100 ⁇ g, or 2 to 10 ⁇ g, per 10 5 NK cells.
  • an NK cell may contain 1 to 10 3 or 10 to 10 2 exogenous mitochondria.
  • the number of exogenous mitochondria may be such that approximately 1, 10, 100, or 500 exogenous mitochondria are contained in one NK cell.
  • the number of exogenous mitochondria contained in NK cells can be regulated by controlling an amount of exogenous mitochondria to be introduced into NK cells.
  • Each subject NK cell may contain a different number of exogenous mitochondria.
  • the NK cells may be derived from a mammal or a human.
  • the NK cells may be obtained from a subject intended to receive NK cell therapy.
  • the NK cells may be directly isolated from the blood of the subject and used, or may be obtained by differentiating immature NK cells or stem cells obtained from the subject and used.
  • the exogenous mitochondria and the NK cells may be mixed and then the mixture may be subjected to centrifugation so that the mitochondria are delivered efficiently into the NK cells.
  • a condition at the time of performing centrifugation can be appropriately regulated to efficiently introduce the mitochondria without damaging the cells.
  • the centrifugation may be performed at room temperature, and a temperature condition can be appropriately selected for cell stability.
  • the NK cells and the exogenous mitochondria may be mixed in the presence of a surfactant so as to increase membrane permeability of the exogenous mitochondria into the NK cells, thereby increasing introduction efficiency of the exogenous mitochondria.
  • the centrifugation may be performed at 100 ⁇ g, 300 ⁇ g, 500 ⁇ g, 800 ⁇ g, 1,000 ⁇ g, 1,200 ⁇ g, 1,500 ⁇ g, 1,800 ⁇ g, 2,000 ⁇ g, 2,400 ⁇ g, 3,000 ⁇ g, 5,000 ⁇ g, or 10,000 ⁇ g.
  • a centrifugation time may be 0.1 minutes to 60 minutes, but is not limited thereto. Specifically, the centrifugation time may be 1 minute, 2 minutes, 3 minutes, 5 minutes, 10 minutes, 20 minutes, or 30 minutes.
  • the centrifugation may be performed at a temperature of 0° C. to 40° C., 20° C. to 38° C., or 30° C. to 37° C.
  • the mitochondria can be delivered into the NK cells with high efficiency while causing less damage to the NK cells.
  • a surfactant can be used to enhance cell membrane permeability of the exogenous mitochondria into the NK cells.
  • a time point at which the surfactant is added may be before, during, or after mixing of the NK cells with the exogenous mitochondria.
  • the NK cells may be incubated for a certain period of time in order to increase the cell membrane permeability of the exogenous mitochondria into the NK cells.
  • An incubation time may be 0.1 to 60 minutes. Specifically, the incubation time may be 1 minute, 5 minutes, 10 minutes, 20 minutes, or 30 minutes, but is not limited thereto.
  • the surfactant is preferably a nonionic surfactant, and may be a poloxamer.
  • the poloxamer is a triblock copolymer composed of a central hydrophobic chain of polyoxypropylene flanked by two hydrophilic chains of polyoxyethylene.
  • the surfactant in the mixture may have a concentration of 1 to 100 mg/ml, 3 to 80 mg/ml, or 5 to 40 mg/ml, and may be preferably 10 to 30 mg/ml.
  • the method may further comprise a step of incubating the mixture under a predetermined time and temperature condition.
  • the incubation may be performed at a temperature of 0° C. to 40° C., 20° C. to 38° C., or 30° C. to 37° C.
  • the incubation may be performed for 0.1 to 4 hours, 0.5 to 3.8 hours, or 0.8 to 3.5 hours.
  • the incubation may be performed for a predetermined time after centrifugation is performed so as to deliver the exogenous mitochondria into the NK cells.
  • the incubation time can be appropriately selected depending on a cell type and an amount of mitochondria.
  • a pharmaceutical composition for treating cancer or an infectious disease comprising, as an active ingredient, an NK cell that contains the exogenous mitochondria.
  • the cancer may be any one selected from the group consisting of gastric cancer, liver cancer, lung cancer, colorectal cancer, breast cancer, prostate cancer, ovarian cancer, pancreatic cancer, cervical cancer, thyroid cancer, laryngeal cancer, acute myelogenous leukemia, brain tumor, neuroblastoma, retinoblastoma, head and neck cancer, salivary gland cancer, and lymphoma.
  • the infectious disease may be any one selected from the group consisting of hepatitis B, hepatitis C, human papilloma virus (HPV) infection, cytomegalovirus infection, viral respiratory disease, and influenza.
  • the pharmaceutical composition may be made into a preparation in liquid or frozen form. Even in a case of being thawed again after freezing, the pharmaceutical composition does not exhibit impaired cellular function and can maintain high cell viability and cell-killing ability. Therefore, the pharmaceutical composition can be easily stored and supplied in a liquid- or frozen-stored form without additional processing.
  • a method for preventing or treating a disease comprising a step of administering, to a subject, a pharmaceutical composition which contains, as an active ingredient, NK cells exogenous mitochondria.
  • Such a method comprises a step of administering an effective amount of the NK cell of the invention to a subject having a disease or a subject suspected of having a disease.
  • a cell into which exogenous mitochondria has been introduced can be administered to a subject, preferably a mammal, as a therapeutic preparation.
  • the cell can be administered by an intravenous or subcutaneous route.
  • the composition of the present invention is provided parenterally such as by intravenous, subcutaneous, ophthalmic, intraperitoneal, or intramuscular route
  • the composition is preferably in an aqueous form, or it is preferable that the composition includes a physiologically applicable body fluid, suspension, or solution.
  • a carrier or vehicle is physiologically acceptable, and thus can be added to the composition and delivered to a patient.
  • a carrier or vehicle does not adversely affect electrolyte of the patient. Therefore, physiological saline can be generally used as a carrier for preparations.
  • the method for preventing or treating a disease using the cell of the present invention may also comprise administering another drug or physiologically active substance having an effect of preventing or treating the disease, in combination with the cell of the present invention.
  • a route, a time, and a dose for the combined administration can be determined depending on the type of disease, the patient's disease state, the purpose of treatment or prevention, and the other drug or physiologically active substance used in combination.
  • the disease may be cancer or an infectious disease.
  • the cancer may be selected from the group consisting of gastric cancer, liver cancer, lung cancer, colorectal cancer, breast cancer, prostate cancer, ovarian cancer, pancreatic cancer, cervical cancer, thyroid cancer, laryngeal cancer, acute myelogenous leukemia, brain tumor, neuroblastoma, retinoblastoma, head and neck cancer, salivary gland cancer, and lymphoma.
  • the infectious disease may be any one selected from the group consisting of hepatitis B, hepatitis C, human papilloma virus (HPV) infection, cytomegalovirus infection, viral respiratory disease, and influenza.
  • HPV human papilloma virus
  • PBMC comprising an exogenous mitochondria.
  • peripheral blood mononuclear cell refers to a cell with a spherical nucleus present in the peripheral blood, which is referred to as peripheral blood monocyte or PBMC.
  • PBMCs may include immune cells such as B cells, T cells, macrophages, dendritic cells, and NK cells.
  • the PBMC can be obtained through the blood of a subject.
  • the exogenous mitochondria can be obtained from tissues or cells of the subject as described above.
  • the PBMC comprising exogenous mitochondria may be obtained by introducing mitochondria in an amount of 0.01 to 500 ⁇ g, 0.1 to 450 ⁇ g, 0.5 to 300 ⁇ g, 1 to 100 ⁇ g, or 2 to 10 ⁇ g, per 10 5 PBMCs.
  • the exogenous mitochondria may be contained in an amount of 1 to 10 3 or 10 to 100, per one PBMC.
  • a method of introducing the exogenous mitochondria into the PBMC can be carried out through centrifugation as described above.
  • a pharmaceutical composition for treating cancer or an infectious disease comprising, as an active ingredient, the PBMC that contains the exogenous mitochondria.
  • the cancer may be selected from the group consisting of gastric cancer, liver cancer, lung cancer, colorectal cancer, breast cancer, prostate cancer, ovarian cancer, pancreatic cancer, cervical cancer, thyroid cancer, laryngeal cancer, acute myelogenous leukemia, brain tumor, neuroblastoma, retinoblastoma, head and neck cancer, salivary gland cancer, and lymphoma.
  • the infectious disease may be any one selected from the group consisting of hepatitis B, hepatitis C, human papilloma virus (HPV) infection, cytomegalovirus infection, viral respiratory disease, and influenza.
  • WRL-68 Human normal hepatocytes (WRL-68) (CRL 1458, ATCC) were seeded in Dulbecco Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS; Gibco), 100 ⁇ g/ml of streptomycin, and 100 U/ml of ampicillin and cultured for 72 hours. After completion of the culture, the cells were washed twice with Dulbecco Phosphate Buffered Saline (DPBS; Gibco). The washed cells were treated with 0.25% trypsin-EDTA (TE; Gibco) to obtain cells. For the obtained cells, in order to extract mitochondria, a hemocytometer was used to measure the number of cells, and cells in an amount of about 3 ⁇ 10 6 cells/ml were collected.
  • DMEM Dulbecco Modified Eagle Medium
  • FBS fetal bovine serum
  • TE trypsin-EDTA
  • the cell line was subjected to primary centrifugation at a temperature of about 4° C. for 10 minutes with a speed of 350 ⁇ g.
  • the resulting pellet was collected, and resuspended and homogenized in a buffer solution for 10 to 15 minutes.
  • a composition containing the pellet was subjected to secondary centrifugation at a temperature of about 4° C. for 3 minutes with a speed of 1,100 ⁇ g, to obtain supernatant.
  • the supernatant was subjected to tertiary centrifugation at a temperature of about 4° C. for 15 minutes with a speed of 12,000 ⁇ g, to isolate mitochondria from the cell line.
  • the isolated mitochondria were injected, in an amount of 1 ⁇ 10 5 , into a test tube containing separate human NK cells (NK92mi) (CRL2408; ATCC), and centrifugation was performed at a temperature of about 4° C. for 15 minutes with a speed of 2,500 ⁇ g. After removal of supernatant, washing with PBS was performed and centrifugation was performed at a temperature of about 4° C. for 5 minutes. Washing was performed twice under the same condition.
  • the isolated mitochondria were delivered at weights of 0.05, 0.05, 0.5, and 5 ⁇ g, per 1 ⁇ 10 5 recipient cells.
  • a DNA purification kit (NucleoSpin; MACHEREY-NAGEL GmbH & Co. KG) was used to extract the entire gene from the NK cells collected in Example 1.
  • the extracted DNA was respectively mixed with WRL-68 mitochondria-specific identification primers (F: 5′-CTA TTC TCT GTT CTT TCA TGG-3′ (SEQ ID NO: 1), R: 5′-AAG TAT TTA TGG TAC CGT ACG-3′ (SEQ ID NO: 2)).
  • a PCR reaction was performed to obtain amplified DNA.
  • electrophoresis on a 1.5% agarose gel was performed, and then staining with Loading Star (DYNEBIO INC., Seongnam, Korea) was performed.
  • Loading Star DYNEBIO INC., Seongnam, Korea
  • a UV-spectrometer (Chemi-Doc XRS; Bio-Rad Laboratories, Inc., Hercules, Calif., USA) was used to identify amplified DNA bands.
  • GAPDH was selected as a house keeping gene.
  • primers F-5′-GGA AGG TGA AGG TCG GAG-3′ (SEQ ID NO: 3), R-5′-GGC AAC AAT ATC CAC TTT ACC-3′ (SEQ ID NO: 4) capable of amplifying GADPH were used. This result is illustrated in FIG. 1 .
  • FIG. 1 indicated that an amount of the exogenous mitochondria delivered into the NK cells is increased as amounts (0.005, 0.05, 0.5, and 5 ⁇ g) of the mitochondria to be mixed with the NK cells are increased.
  • Fluorescence-activated cell sorter analysis was performed to identify whether hepatocyte-derived mitochondria had been delivered into NK cells.
  • the mitochondria isolated from human normal hepatocytes were treated with 500 nM Green mitotracker (Thermo Fisher Scientific, Waltham, USA). The resultant was allowed to react for 10 minutes in a 5% CO 2 incubator at 37° C., and washed.
  • the fluorescence-labeled mitochondria derived from hepatocytes were delivered into immune cells using a centrifugation method, and then the immune cells were resuspended in 1 mL of PBS. Then, the mitochondrial delivery was identified and analyzed using the FACS Calibur flow cytometer (BDBiosciences, San Jose, Calif., USA). The result is illustrated in FIG. 2 .
  • the NK cells can be distinguished depending on amounts (0.005, 0.05, 0.5, and 5 ⁇ g) of the mitochondria delivered into the NK cells.
  • NK92mi human NK cells
  • mitochondria of the NK cells were treated with 500 nM Green mitotracker (Thermo Fisher Scientific, Waltham, USA), and allowed to react for 10 minutes in a 5% CO 2 incubator at 37° C.
  • the isolated mitochondria of the hepatocytes were treated with 500 nM red mitotracker.
  • the resultant was allowed to react for 10 minutes in a 5% CO 2 incubator at 37° C., and then delivered into NK cells. After 5 ⁇ g of the mitochondria was delivered, the resultant was seeded in a 24-well plate and incubated in a 5% CO 2 incubator at 37° C. Then, fluorescence microscopy was used to identify intracellular delivery within 24 hours.
  • a DAPI reagent for nuclear staining was used as a control staining reagent. The results are illustrated in FIG. 3 .
  • NK92mi human NK cells collected in Example 1, into which exogenous mitochondria had been introduced, in order to identify expression of CD107a due to degranulation which is an indicator for NK cell activity, the human NK cells and target cells (K562) were mixed at a ratio of 10:1, and then the mixture was treated with fluorescent material-conjugated anti-CD107a. The resultant was co-incubated for 4 hours. After the co-incubation, the resultant was treated with anti-CD56 for surface staining and allowed to react for 30 minutes. Then, fluorescence-activated cell sorter (FACS) analysis was performed. The results are illustrated in FIGS. 4A and 4B .
  • FACS fluorescence-activated cell sorter
  • the collected NK cells were mixed, at 10:1, with target cells (K562) labeled with green fluorescence staining (CFSE; Invitrogen), and then the mixture was co-incubated for 4 hours in an incubator with a condition of 5% CO 2 at 37° C. After the co-incubation, in order to analyze the target cells killed by the NK cells, the resultant was treated with red fluorescence staining (7-AAD; Invitrogen), and then allowed to react for 10 minutes. Fluorescence intensity of the killed target cells was analyzed with fluorescence-activated cell sorter (FACS). The result is illustrated in FIG. 5 .
  • FACS fluorescence-activated cell sorter
  • Placenta (provided by CHA bundang medical center, IRB No. 1044308-201511-BR-022-02)-derived mesenchymal stem cells were seeded in Alpha-Minimum Essential Medium (Alpha-MEM) supplemented with 10% fetal bovine serum (FBS; Gibco), 100 ⁇ g/ml of streptomycin, and 100 U/ml of ampicillin, and cultured for 72 hours.
  • Alpha-MEM Alpha-Minimum Essential Medium
  • FBS fetal bovine serum
  • streptomycin 100 ⁇ g/ml of streptomycin
  • ampicillin 100 U/ml of ampicillin
  • the cells were washed twice with Dulbecco Phosphate Buffered Saline (DPBS; Gibco).
  • DPBS Dulbecco Phosphate Buffered Saline
  • the washed cells were treated with 0.25% trypsin-EDTA (TE; Gibco) to obtain cells.
  • TE trypsin-EDTA
  • a hemocytometer was used to measure the number of cells, and cells in an amount of about 2 ⁇ 10 7 cells/ml were collected.
  • the cell line was subjected to primary centrifugation at a temperature of about 4° C. for 10 minutes with a speed of 350 ⁇ g.
  • the resulting pellet was collected, and resuspended and homogenized in a buffer solution for 10 to 15 minutes.
  • a composition containing the pellet was subjected to secondary centrifugation at a temperature of about 4° C. for 3 minutes with a speed of 1,100 ⁇ g, to obtain supernatant.
  • the supernatant was subjected to tertiary centrifugation at a temperature of about 4° C. for 15 minutes with a speed of 12,000 ⁇ g, to isolate mitochondria from the cell line.
  • the isolated mitochondria were injected, in an amount of 1 ⁇ 10 5 , into a test tube containing separate human NK cells (NK92mi) (CRL2408; ATCC), and centrifugation was performed at a temperature of about 4° C. for 15 minutes with a speed of 2,500 ⁇ g. After removal of supernatant, washing with PBS was performed and centrifugation was performed at a temperature of about 4° C. for 5 minutes. Washing was performed twice under the same condition.
  • the isolated mitochondria were delivered at weights of 0.3, 1, 3, 5, and 10 ⁇ g, per 1 ⁇ 10 5 recipient cells.
  • Fluorescence-activated cell sorter analysis was performed to identify whether mitochondria derived from umbilical cord-derived mesenchymal stem cells had been delivered into NK cells. Mitochondria isolated from the umbilical cord-derived mesenchymal stem cells were treated with 500 nM Red mitotracker (Thermo Fisher Scientific, Waltham, USA). The resultant was allowed to react for 30 minutes in a 5% CO 2 incubator at 37° C. and washed. The fluorescence-labeled mitochondria of the umbilical cord-derived mesenchymal stem cells were delivered into immune cells using a centrifugation method, and then the cells were resuspended in 1 mL of PBS. Then, the mitochondrial delivery was identified and analyzed using the FACS Calibur flow cytometer (BDBiosciences, San Jose, Calif., USA). The results are illustrated in FIG. 6 .
  • FACS Fluorescence-activated cell sorter
  • the NK cells can be distinguished with amounts (0.3, 1, 3, 5, and 10 ⁇ g) of the UC-MSC-derived mitochondria which have been delivered into the NK cells.
  • the collected NK cells were mixed, at 10:1, with target cells (K562) labeled with green fluorescence staining (CFSE; Invitrogen), and then the mixture was co-incubated for 4 hours in an incubator with a condition of 5% CO 2 at 37° C. After the co-incubation, in order to analyze the target cells killed by the NK cells, the resultant was treated with red fluorescence staining (7-AAD; Invitrogen), and allowed to react for 10 minutes. Fluorescence intensity of the killed target cells was analyzed with fluorescence-activated cell sorter (FACS). The results are illustrated in FIG. 7 .
  • mice 6- to 8-week-old male NOD.cg-Prkdcscid IL2rgtm1Sug/JicKoat mice were purchased from Koatech Co., Ltd. (Gyeonggi-do, Korea). The purchased mice were subjected to an adaptation period in a clean zone of the experimental animal center at the CHA University, and then an experiment was conducted. During the adaptation period, the environment in which the mice are kept had day and night at a 12-hour interval, and was maintained at a room temperature of 23 ⁇ 2° C. and a humidity of 40% to 60%. The mice were subjected to such an adaptation period for 7 days, and then put into the experiment. The mice thus prepared were administered, via the tail vein (intravenous (i.v.) injection), K562 cells in an amount of 2 ⁇ 10 5 cells/100 ⁇ l, so that an acute myelogenous leukemia model was produced.
  • i.v. intravenous
  • mice in which acute myelogenous leukemia had been induced were administered, via the tail vein (intravenous (i.v.) injection), the NK cells (NK92mi) prepared according to Example 7 in an amount of 2 ⁇ 10 6 cells/100 ⁇ l, so that an experimental group was produced.
  • a control group was produced by administration of normal NK cells (NK92mi), into which the mitochondria had not been delivered, in an amount of 2 ⁇ 10 6 cells/100 ⁇ l.
  • Body weight change and survival rate of the experimental group and the control group were analyzed for 24 days from a time point at which the acute myelogenous leukemia cell line (K562) had been administered. The results are illustrated in FIGS. 8A to 8C .
  • the blood of the experimental group and the control group to which an experiment had been conducted according to Example 10 was collected, and then centrifuged at 12,000 ⁇ g for 15 minutes to isolate the serum.
  • the isolated serum was analyzed for expression distribution of p53 and c-Myc using the Western Blot Kit (WB; Bio-Rad Laboratories, Inc.). The results are illustrated in FIG. 9 .
  • the peripheral blood of the inventor which had been taken by a clinician was treated with Ficoll-Paque (Amersham Biosciences) at 1:1 in a Falcon tube, and then the resultant was subjected to centrifugation at 400 ⁇ g for 35 minutes, to collect a PBMC pellet.
  • the collected PBMCs were washed twice with PBS.
  • the human hepatocyte (WRL-68)-derived mitochondria which had been isolated according to Example 1 were delivered into the PBMCs at weights of 0.05, 0.05, 0.5, and 5 ⁇ g, per 1 ⁇ 10 5 recipient cells.
  • the collected PBMCs were mixed, at 10:1, with target cells (K562) labeled with green fluorescence staining (CFSE; Invitrogen), and then the mixture was co-incubated for 4 hours in an incubator with a condition of 5% CO 2 at a temperature of 37° C.
  • target cells K562 labeled with green fluorescence staining (CFSE; Invitrogen)
  • CFSE green fluorescence staining
  • the resultant was treated with red fluorescence staining (7-AAD; Invitrogen), and then allowed to react for 10 minutes. Fluorescence intensity of the killed target cells was analyzed with fluorescence-activated cell sorter (FACS). The result is illustrated in FIG. 10 .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Developmental Biology & Embryology (AREA)
  • Oncology (AREA)
  • Mycology (AREA)
  • Virology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US16/349,310 2016-11-14 2017-11-14 Natural killer cell containing exogenous mitochondrium and pharmaceutical composition comprising same Active 2038-11-09 US11278569B2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20160151411 2016-11-14
KR10-20160151411 2016-11-14
KR10-2016-0151411 2016-11-14
PCT/KR2017/012883 WO2018088875A2 (fr) 2016-11-14 2017-11-14 Cellule tueuse naturelle contenant une mitochondrie exogène et composition pharmaceutique la comprenant

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2017/012883 A-371-Of-International WO2018088875A2 (fr) 2016-11-14 2017-11-14 Cellule tueuse naturelle contenant une mitochondrie exogène et composition pharmaceutique la comprenant

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/483,193 Continuation US20220000930A1 (en) 2016-11-14 2021-09-23 Natural killer cell containing exogenous mitochondrium and pharmaceutical composition comprising same

Publications (2)

Publication Number Publication Date
US20190269731A1 US20190269731A1 (en) 2019-09-05
US11278569B2 true US11278569B2 (en) 2022-03-22

Family

ID=61977886

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/349,310 Active 2038-11-09 US11278569B2 (en) 2016-11-14 2017-11-14 Natural killer cell containing exogenous mitochondrium and pharmaceutical composition comprising same
US17/483,193 Pending US20220000930A1 (en) 2016-11-14 2021-09-23 Natural killer cell containing exogenous mitochondrium and pharmaceutical composition comprising same

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/483,193 Pending US20220000930A1 (en) 2016-11-14 2021-09-23 Natural killer cell containing exogenous mitochondrium and pharmaceutical composition comprising same

Country Status (7)

Country Link
US (2) US11278569B2 (fr)
EP (1) EP3540048A4 (fr)
JP (1) JP7074358B2 (fr)
KR (2) KR101846464B1 (fr)
CN (1) CN109983120A (fr)
RU (1) RU2021134402A (fr)
WO (1) WO2018088875A2 (fr)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20200071338A (ko) 2018-12-11 2020-06-19 주식회사 스마트셀랩 Nk 세포 활성 조절을 통한 비만의 예방 또는 치료 방법
CN114786685A (zh) * 2019-07-09 2022-07-22 唐凌峰 一种基于线粒体的药物递送系统及其用途
CN116096861A (zh) * 2020-04-03 2023-05-09 赛立维公司 过继性细胞转移的增强
KR102273163B1 (ko) 2020-09-10 2021-07-05 주식회사 파이안바이오테크놀로지 혈소판 유래 미토콘드리아의 수득 방법 및 이의 용도
KR102290596B1 (ko) 2020-09-10 2021-08-19 주식회사 파이안바이오테크놀로지 분리된 미토콘드리아를 포함하는 주사용 조성물 및 이의 용도
WO2023060212A1 (fr) * 2021-10-06 2023-04-13 Cellvie Inc. Amélioration du transfert cellulaire adoptif par la promotion d'une population supérieure de cellules immunitaires adaptatives
CN114699430A (zh) * 2022-04-12 2022-07-05 重庆理工大学 线粒体及其在胰腺炎中的应用和应用方法

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130034527A1 (en) 2010-10-28 2013-02-07 Searete Llc Mitochondrial enhancement of cells
WO2013094988A1 (fr) 2011-12-22 2013-06-27 Mogam Biotechnology Research Institute Procédé pour produire des cellules tueuses naturelles, cellules tueuses naturelles produites de cette façon, et composition pour traiter des cancers et des maladies infectieuses contenant les mêmes
WO2015067089A1 (fr) 2013-11-08 2015-05-14 中国科学院广州生物医药与健康研究院 Procédé pour introduire des mitochondries exogènes dans des cellules de mammifère
KR20150063374A (ko) 2012-08-13 2015-06-09 안트로제네시스 코포레이션 자연 살해 세포 및 그의 용도
WO2016008937A1 (fr) 2014-07-16 2016-01-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de transfert intercellulaire de mitochondries isolées dans des cellules receveuses
WO2016135723A1 (fr) 2015-02-26 2016-09-01 Minovia Therapeutics Ltd. Cellules mammifères enrichies avec des mitochondries fonctionnelles

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130034527A1 (en) 2010-10-28 2013-02-07 Searete Llc Mitochondrial enhancement of cells
WO2013094988A1 (fr) 2011-12-22 2013-06-27 Mogam Biotechnology Research Institute Procédé pour produire des cellules tueuses naturelles, cellules tueuses naturelles produites de cette façon, et composition pour traiter des cancers et des maladies infectieuses contenant les mêmes
KR20140123503A (ko) 2011-12-22 2014-10-22 재단법인 목암생명공학연구소 자연살해세포의 제조방법, 이러한 방법에 의해 제조된 자연살해세포 및 이를 포함하는 종양 및 감염성 질환 치료용 조성물
KR20150063374A (ko) 2012-08-13 2015-06-09 안트로제네시스 코포레이션 자연 살해 세포 및 그의 용도
US20150225697A1 (en) 2012-08-13 2015-08-13 Anthrogenesis Corporation Natural killer cells and uses thereof
WO2015067089A1 (fr) 2013-11-08 2015-05-14 中国科学院广州生物医药与健康研究院 Procédé pour introduire des mitochondries exogènes dans des cellules de mammifère
WO2016008937A1 (fr) 2014-07-16 2016-01-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de transfert intercellulaire de mitochondries isolées dans des cellules receveuses
WO2016135723A1 (fr) 2015-02-26 2016-09-01 Minovia Therapeutics Ltd. Cellules mammifères enrichies avec des mitochondries fonctionnelles

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Andrés Caicedo et al., "Artificial Mitochondria Transfer: Current Challenges, Advances, and Future Applications," Stem Cells International, vol. 2017, Jul. 1, 2017, pp. 1-23 (23 pages).
Andrés Caicedo et al., "MitoCeption as a new tool to assess the effects of mesenchymal stem/stromal cell mitochondria on cancer cell metabolism and function", Scientific Reports, vol. 5, No. 1, Mar. 13, 2015, pp. 1-10 (10 pages total).
International Search Report for PCT/KR2017/012883 dated Feb. 14, 2018 [PCT/ISA/210].
Kitani, T. et al.: "Internalization of Isolated Functional Mitochondria: Involvement of Macropinocytosis", Journal of Cellular and Molecular Medicine, vol. 18, No. 8, Jun. 2014 , pp. 1694-1703 (10 pages total).
Korea Office Action for Application No. 10-2017-0151503 dated Mar. 13, 2018.
Mi Jin Kim et al., "Delivery of exogenous mitochondria via centrifugation enhances cellular metabolic function", Scientific Reports, vol. 8, No. 1, Feb. 20, 2018, pp. 2-13 ( 13 pages total).
Tomoya Kitani, et al., "Internalization of isolated functional mitochondria: involvement of macropinocytosis", Journal of Cellular and Molecular Medicine, 2014, pp. 1694-1703, vol. 18, No. 8.
Voigt J. et al., "Human natural killer cells acting as phagocytes against Candida albicans and mounting an inflammatory response that modulates neutrophil antifungal activity," JID, 2014, vol. 209, No. 4, pp. 616-626, p. 620 left col. bottom para, (11 pages total).

Also Published As

Publication number Publication date
WO2018088875A3 (fr) 2018-08-09
EP3540048A4 (fr) 2020-06-24
EP3540048A2 (fr) 2019-09-18
US20190269731A1 (en) 2019-09-05
KR20180054523A (ko) 2018-05-24
RU2019118070A (ru) 2020-12-14
JP2019534029A (ja) 2019-11-28
WO2018088875A2 (fr) 2018-05-17
JP7074358B2 (ja) 2022-05-24
KR101846464B1 (ko) 2018-04-09
US20220000930A1 (en) 2022-01-06
RU2021134402A (ru) 2022-01-14
RU2019118070A3 (fr) 2021-08-13
CN109983120A (zh) 2019-07-05

Similar Documents

Publication Publication Date Title
US20220000930A1 (en) Natural killer cell containing exogenous mitochondrium and pharmaceutical composition comprising same
Ge et al. Infusion of mesenchymal stem cells and rapamycin synergize to attenuate alloimmune responses and promote cardiac allograft tolerance
AU2015354941B2 (en) Method for culturing natural killer cells using T cells
KR20170121178A (ko) 보편적인 살해 t-세포
JP2017532353A (ja) ナチュラルキラー細胞のための方法及び組成物
CA2649874C (fr) Cellules progenitrices immuno privilegiees et modulatrices
US20220275334A1 (en) Nk cell compositions and preparations for immunotherapy and methods for their production
CN110846281B (zh) 一种基于外泌体的抗肿瘤疫苗
JP2022065022A (ja) 改変ヒト初代血液樹状細胞株を生成するための方法
JP2018531022A6 (ja) 改変ヒト初代血液樹状細胞株を生成するための方法
CA3149478A1 (fr) Compositions cellulaires comprenant des vecteurs viraux et procedes de traitement
Plantinga et al. Clinical grade production of wilms’ tumor-1 loaded cord blood-derived dendritic cells to prevent relapse in pediatric AML after cord blood transplantation
JP2017520582A (ja) 関節リウマチ治療のための間葉系間質細胞
Shin et al. Cytokine engineered NK-92 therapy to improve persistence and anti-tumor activity
Qin et al. Anti-glioma response of autologous T cells stimulated by autologous dendritic cells electrofused with CD133+ or CD133− glioma cells
RU2727540C1 (ru) Применение мембранных везикул мультипотентных стромальных клеток, индуцированных цитохалазином В, для восстановления и повышения митохондриальной функции
JP2023513632A (ja) 血小板由来ミトコンドリア治療及び多能性細胞の生成方法
KR102032384B1 (ko) 제대혈 단핵세포에서의 자연살해세포의 제조 방법
RU2777371C2 (ru) Клетка-естественный киллер, содержащая экзогенную митохондрию, и фармацевтическая композиция, включающая такую клетку
Zhang et al. Inhibition of cancer cell immune evasion by combined application of cytotoxic T-lymphocytes and natural killer cells
US10022402B2 (en) Allogenic mesendritic vector for ovarian cancer
Kapranov et al. Co-culturing of multipotent mesenchymal stromal cells with autological and allogenic lymphocytes
KR20220091426A (ko) 혈액 유래 물질 및 면역세포를 포함하는 암 또는 면역질환의 예방 또는 치료용 약학조성물
Stagg et al. Properties of mesenchymal stem cells to consider for cancer cell therapy
CN117363578A (zh) 一种延缓car-t细胞耗竭的细胞生产方法

Legal Events

Date Code Title Description
AS Assignment

Owner name: PAEAN BIOTECHNOLOGY INC., KOREA, REPUBLIC OF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HAN, KYUBOEM;LEE, YOUNGJUN;REEL/FRAME:049161/0574

Effective date: 20190410

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: AWAITING TC RESP., ISSUE FEE NOT PAID

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: AWAITING TC RESP., ISSUE FEE NOT PAID

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STCF Information on status: patent grant

Free format text: PATENTED CASE